WO2001087306A2 - Compositions and methods for the treatment of colorectal cancer - Google Patents

Compositions and methods for the treatment of colorectal cancer Download PDF

Info

Publication number
WO2001087306A2
WO2001087306A2 PCT/US2001/015326 US0115326W WO0187306A2 WO 2001087306 A2 WO2001087306 A2 WO 2001087306A2 US 0115326 W US0115326 W US 0115326W WO 0187306 A2 WO0187306 A2 WO 0187306A2
Authority
WO
WIPO (PCT)
Prior art keywords
thalidomide
irinotecan
solvate
clathrate
hydrate
Prior art date
Application number
PCT/US2001/015326
Other languages
English (en)
French (fr)
Other versions
WO2001087306A8 (en
Inventor
Rangaswamy Govindarajan
Andrew Zeitlin
Original Assignee
Celgene Corp.
Board Of Trustees Of The University Of Arkansas
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Celgene Corp., Board Of Trustees Of The University Of Arkansas filed Critical Celgene Corp.
Priority to NZ522766A priority Critical patent/NZ522766A/en
Priority to DE60118547T priority patent/DE60118547T2/de
Priority to AU2001261473A priority patent/AU2001261473B2/en
Priority to EP01935372A priority patent/EP1286671B1/en
Priority to CA2408710A priority patent/CA2408710C/en
Priority to AU6147301A priority patent/AU6147301A/xx
Priority to JP2001583774A priority patent/JP4679028B2/ja
Publication of WO2001087306A2 publication Critical patent/WO2001087306A2/en
Publication of WO2001087306A8 publication Critical patent/WO2001087306A8/en
Priority to HK03106060A priority patent/HK1053975A1/xx

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/724Cyclodextrins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to pharmaceutical compositions comprising thalidomide and irinotecan, to methods of treating colorectal cancer, and to methods of reducing or avoiding adverse effects of irinotecan.
  • Cancer is characterized primarily by an increase in the number of abnormal cells derived from a given normal tissue, invasion of adjacent tissues by these abnormal cells, or lymphatic or blood-borne spread of malignant cells to regional lymph nodes and to distant sites (metastasis).
  • Clinical data and molecular biologic studies indicate that cancer is a multistep process that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia.
  • Hyperplasia is a form of controlled cell proliferation involving an increase in cell number in a tissue or organ, without significant alteration in structure or function.
  • endometrial hyperplasia often precedes endometrial cancer.
  • Metaplasia is a form of controlled cell growth in which one type of adult or fully differentiated cell substitutes for another type of adult cell. Metaplasia can occur in epithelial or connective tissue cells.
  • Atypical metaplasia involves a somewhat disorderly metaplastic epithelium.
  • Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia; it is the most disorderly form of non-neoplastic cell growth, involving a loss in individual cell uniformity and in the architectural orientation of cells.
  • Dysplastic cells often have abnormally large, deeply stained nuclei, and exhibit pleomo ⁇ hism.
  • Dysplasia characteristically occurs where there exists chronic irritation or inflammation, and is often found in the cervix, respiratory passages, oral cavity, and gall bladder.
  • the neoplastic lesion may evolve clonally and develop an increasing capacity for invasion, growth, metastasis, and heterogeneity, especially under conditions in which the neoplastic cells escape the host's immune surveillance.
  • cancers of the colon and 5 rectum spread in at least five ways: directed extension through the bowel wall; hematogenous metastases; regional lymph node metastases; perineural spread; and intraluminal metastases. Id.
  • 5-fluorouracil 5-fluorouracil
  • methyl-CCNU 5-fluorouracil
  • CPT-1 1 (irinotecan), the only drug currently approved in the US for second line treatment of colorectal cancer.
  • Overall response rates for CPT-11 are less than 20 percent, however, and complete response is achieved in less than one percent.
  • the median duration of response is only 6.4 months. See, e.g., Rothenberg ML, et al, Cancer 85(4):786-795 (1999); and Cunningham, D, et al, The Lancet 352(9138): 1413-1418 (1998).
  • CPT-11 The main dose-limiting factor of CPT-11 is severe and frequent gastrointestinal (GI) toxicity, particularly diarrhea, which may be early onset (within 24 hours of drug administration) or late onset (more than 24 hours after administration). Early onset diarrhea is uncommon, cholinergic in nature, and responds to atropine. In contrast, debilitating (Grade 3-4) late onset diarrhea occurs in 30-40 percent of patients and adversely affects GI toxicity.
  • GI gastrointestinal
  • diarrhea which may be early onset (within 24 hours of drug administration) or late onset (more than 24 hours after administration).
  • Early onset diarrhea is uncommon, cholinergic in nature, and responds to atropine.
  • debilitating (Grade 3-4) late onset diarrhea occurs in 30-40 percent of patients and adversely affects
  • Irinotecan also referred to as CPT-11 and chemically named (4S)-4,1 l-diethyl-4- hydroxy-9-[(4-piperidino-piperidino)carbonyl-oxy]lH-pyranol-[3',4':6,7]indolizinol[l,2- b]quinoline-3,14-(4H,12H)dione, is described in U.S. Patent No. 4,604,463.
  • the hydrochloride trihydrate of irinotecan is sold under the tradename CAMPTOSAR ® , and is indicated in the United States for the treatment of patients with metastatic carcinoma of the
  • Physicians' Desk Reference, 2412-2418 (54 th ed., 2000). It has also recently been approved in the United States as a first-line therapy to treat patients with metastic colorectal cancer in combination with 5-fluorouracil and leucovorin. Irinotecan has also reportedly been used to treat other cancers, such as malignant gliomas and NSCLC. See, e.g., Avgeropoulos, N.G., and Batchelor, T.T., The Oncologist 4:209-224 (1999).
  • Topoisomerases are enzymes that catalyze the relaxation of negatively supercoiled deoxyribonucleic acid (DNA). The process they catalyze is believed to comprise three steps: cleavage of one or both strands of a supercoiled DNA; passage of a segment of DNA through the break that is formed; and resealing of the break.
  • Type I topoisomerases cleave one strand of DNA; type II topoisomerases cleave both strands. Stryer, L., Biochemistry 662-663 (3 rd ed., 1988).
  • topoisomerase inhibitors have thus shown promise in the treatment of various types of cancer.
  • Irinotecan has numerous adverse effects, examples of which include, but are not limited to, early and late-forming diarrhea, nausea, vomiting, anorexia, constipation, flatulence, leukopenia, anemia, neutropenia, asthenia, abdominal cramping, fever, pain, loss of body weight, dehydration, alopecia, dyspnea, insomnia, and dizziness. See, e.g., Physicians' Desk Reference, 2415 (54 lh ed., 2000). The mechanisms by which these undesired effects occur are not well understood, but are believed to be different. In particular, the early and late- forms of diarrhea typically experienced by patients are reportedly mediated by different mechanisms. Id.
  • irinotecan limits the amount of irinotecan that can be administered to patients.
  • the effectiveness of irinotecan is consequently limited not only by its ability to inhibit topoisomerase activity, but also by the severity and nature of its adverse effects.
  • Attempts have been made to alleviate adverse effects associated with irinotecan.
  • loperamide and the combination of loperamide and aceto ⁇ han have reportedly been administered to patients in an effort to reduce delayed-onset diarrhea. Rothenberg, M ., Annals of Oncology 8:837-855 (1997). Unfortunately, these attempts met with limited success. Id.
  • Thalidomide is a racemic compound sold under the tradename THALOMID ® and chemically named ⁇ -(N-phthalimido)glutarimide or 2-(2,6-diox.o-3-piperidinyl)-lH- isoindole-l,3(2H)-dione.
  • Thalidomide was originally developed in the 1950's to treat morning sickness, but due to its tetragenic effects was withdrawn- from use. Thalidomide is now indicated in the United States for the acute treatment of the cutaneous manifestations of erythema nodosum leprosum. Physicians' Desk Reference, 911-916 (54 lh ed., 2000). Because its administration to pregnant women can cause birth defects, the sale of thalidomide is strictly controlled. Id.
  • thalidomide has reportedly been used to treat chronic graft-vs-host disease, rheumatoid arthritis, sarcoidosis, several inflammatory skin diseases, and inflammatory bowel disease. See generally, Koch, H.P., Prog. Med. Chem. 22:165-242 (1985). See also, Moller, D.R., et al, J. Immunol. 159:5157-5161 (1997); Vasiliauskas, E.A., et al, Gastroenterology 1 17:1278-1287 (1999); and Ehrensch, E.D., et al, Gastroenterology 117:1271-1277 (1999). It has further been alleged that thalidomide can be combined with other drugs to treat iscehemia/reperfusion associated with coronary and cerebral occlusion. See U.S. Patent No. 5,643,915, which is incorporated herein by reference.
  • Thalidomide has also reportedly been clinically investigated in the treatment of specific types of cancers. These include refractory multiple myeloma, brain, melanoma, breast, colon, mesothelioma, and renal cell carcinoma. See, e.g., Singhal, S., et al, New England J. Med. 341(21): 1565-1571 (1999); and Marx, G.M., et al, Proc. Am. Soc. Clin. Oncology 18:454a (1999). It has further been reported that thalidomide can be used to prevent the development of chronic cardiomyopathy in rats caused by doxorubicin. Costa, P.T., et al, Blood 92(10:suppl.
  • thalidomide in the treatment of specific cancers include its combination with carboplatin in the treatment of ghoblastoma multiforme. McCann, J., Drug Topics 41-42 (June 21, 1999). Thalidomide has reportedly also been used as an antiemetic during the treatment of astrocytoma. Zwart, D. , Arzneim.-Forsch. 16(12):1688-1689 (1966).
  • thalidomide aids in the treatment of some cancers, its nature remains unclear. See, e.g., Moreira, A.L., et al, J. Expr. Med. 177:1675- 1680 (1993); McHugh, S.M., et al, Clin. Exper. Immunol. 99:160-167 (1995); and Moller, D.R., et al., J. Immunol 159:5157-5161 (1997). It has been reported, however, thai thalidomide is an antiangiogenic agent that can suppress tumor necrosis factor (TNF- ⁇ ) and interleukin 12 (IL-12) production.
  • TNF- ⁇ tumor necrosis factor
  • IL-12 interleukin 12
  • Thalidomide may also affect mechanisms related to epithelial or endothelial function or growth. D'amato M., et al, Proc. Natl. Acad. Sci. 91:4082-4085(1994). Given the great need for an effective and safe treatment of cancer, there continues to be an extensive amount of research on new drugs or ways of improving existing therapies. This invention addresses the need for a safe and effective treatment of colorectal cancer. 3. SUMMARY OF THE INVENTION
  • This invention is directed to pharmaceutical compositions, pharmaceutical dosage forms, kits, and methods of treating primary or metastatic colorectal cancer.
  • a first embodiment of the invention encompasses a method of treating primary and/or metastatic colorectal cancer, which comprises administering to a patient in need of such treatment a therapeutically effective amount of irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, and a therapeutically effective amount of thalidomide, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof.
  • irinotecan is administered parenterally about every three weeks in an amount of from about 1 to about 1000 mg/m 2 , preferably in an amount of from about 25 to about 750 mg/m 2 , more preferably in an amount of from about 50 to about 500 mg/m 2 , and most preferably in an amount of from about 100 to about 350 mg/m 2
  • thalidomide is administered orally and daily in an amount of from about 1 to about 2000 mg, preferably from about 50 to about 1000 mg, more preferably from about 100 to 750 mg, and most preferably from about 200 to about 500 mg.
  • a second embodiment of the invention encompasses a pharmaceutical composition
  • a pharmaceutical composition comprising irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, and thalidomide, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof.
  • a third embodiment of the invention encompasses a dosage form comprising irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, and thalidomide, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof.
  • a fourth embodiment of the invention encompasses a kit for use in the treatment or prevention of colorectal cancer which comprises a parenteral dosage form of irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, and an oral dosage form of thalidomide, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof.
  • This invention encompasses pharmaceutical compositions, pharmaceutical dosage ' forms, kits, and methods of treating primary or metastatic colorectal cancer.
  • This invention is based, in part, on the unique ability of thalidomide to improve the- overall therapeutic profile of irinotecan when used in the treatment of colorectal cancer.
  • the invention consequently encompasses a method of treating or preventing colorectal cancer which comprises the administration of thalidomide, or a derivative, analogue, pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, in combination with irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, to a patient.
  • thalidomide can improve the efficacy of irinotecan at its common or approved dose. Thalidomide can further be used in combination with lower doses of irinotecan to reduce or avoid adverse affects associated with irinotecan while maintaining its efficacy. Thalidomide can also be used to reduce or avoid gastrointestinal toxicity caused by irinotecan.
  • this invention encompasses therapeutic effects that result from an unexpected and unique synergy between thalidomide and irinotecan. One of these therapeutic effects is an increased potency or efficacy of irinotecan; another is a reduced toxicity or increased safety of irinotecan.
  • compositions of the invention include bulk drug compositions useful in the manufacture of pharmaceutical compositions (e.g., impure or non-sterile compositions), pharmaceutical compositions (i.e., compositions that are suitable for administration to a patient), and individual dosage forms.
  • Each of the compositions and dosage forms of the invention comprise at least two of what are referred to herein as "active ingredients. "
  • a first active ingredient is irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof.
  • a second active ingredient is thalidomide, a derivative or analogue of thalidomide, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof.
  • Irinotecan contains a chiral center, and is commercially available as an optically pure compound.
  • the methods and compositions of the invention encompass the use of racemic irinotecan, however, as well as enriched (i.e., uneven) mixtures of its enantiomers.
  • Optically pure enantiomers of irinotecan can be prepared by methods well known in the art. These include, but are not limited to, resolution of chiral salts, asymmetric synthesis, or chiral chromatography. See generally, Beesley, T.E. and Scott, R.P.W., Chiral Chromatography (John Wiley & Sons, New York: 1999); Principles of Asymmetric Synthesis, Gawley, R.E.
  • Thalidomide contains a chiral center, and is sold as a racemate.
  • the methods and compositions of the invention therefore encompass the use of racemic thalidomide as well as optically pure enantiomers of thalidomide.
  • Optically pure enantiomers of thalidomide can be prepared by methods well known in the art.
  • thalidomide examples include, but are not limited to, taglutimide, supidimide, and those disclosed by International Application WO 94/20085, which is inco ⁇ orated herein by reference.
  • thalidomide encompassed by this invention include, but are not limited to, 6- alkyl-2-[3'- or 4'-nitrophthalimido]-glutarimides and 6-alkyl-3-phenylglutarimides. See, e.g., De, A.U., and Pal. D., J. Pharm. Sci. 64(2): 262-266 (1975).
  • This invention encompasses methods of treating cancer of the colon and rectum mammals, and in humans in particular.
  • dosage fo ⁇ ns of the invention can be used in methods of the invention, the active ingredients disclosed herein can be administered separately, in any appropriate form, and by any suitable route.
  • thalidomide can work in combination with irinotecan to more rapidly kill cancer cells, while at the same time reducing gastrointestinal (e.g., diarrhea) and other side effects associated with irinotecan.
  • one embodiment of this invention encompasses methods of treating colorectal cancer, including primary and metastatic colorectal cancer, and preventing metastases of primary colorectal cancer, or preventing further colorectal metastasis. It further encompasses methods of treating patients who have been previously treated for cancer, as well as those who have not previously been treated for cancer. Indeed, the methods and compositions of this invention can be used in first-line and second-line cancer treatments.
  • thalidomide is administered to a patient prior to the observation of intolerance of irinotecan.
  • inventions include methods of increasing the dosage of irinotecan that can be safely and effectively administered to a patient, and methods of varying the dosage cycle used to administer irinotecan to a patient while avoiding dose- limiting toxicities.
  • This invention further encompasses methods of: 1) allowing the completion of chemotherapy in a greater percentage of patients; 2) avoiding deterioration of patients' nutritional status secondary to gastrointestinal toxicity; and 3) improving' the overall quality of patients' life during chemotherapy. 4.1.1. METHODS OF TREATING AND/OR PREVENTING COLORECTAL CANCER
  • the methods of treating and/or preventing colorectal cancer encompassed by this invention comprise administering at least two drugs (also referred to herein as "active ingredients” or “active agents”) to a patient (e.g., a human) suffering, or likely to suffer, from colorectal cancer: 1) irinotecan, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, or prodrug thereof, and; 2) thalidomide, a derivative or analogue of thalidomide, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, or prodrug thereof.
  • the two active ingredients can be administered concurrently, sequentially, and by the same or by different routes of administration.
  • thalidomide can be administered to a patient prior to, during, or after the administration of irinotecan.
  • a prophylactic or therapeutic dose of each active ingredient in the acute or chronic management of colorectal cancer will typically vary with the specific active ingredients, the severity and type of cancer, and the route of administration.
  • the dose, and perhaps the dose frequency may also vary according to age, body w.eight, response, and the past medical history of the patient.
  • Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors by following, for example, dosages reported in the literature and recommended in the Physician 's Desk Reference ® (54 th ed., 2000).
  • irinotecan is administered parenterally about every three weeks in an amount of from about 1 to about 1000 mg/m 2 , preferably in an amount of from about 25 to about 750 mg/m 2 , more preferably in an amount of from about 50 to about 500 mg/m 2 , and most preferably in an amount of from about 100 to about 350 mg/m 2 .
  • thalidomide is administered orally and daily in an amount of from about 1 to about 2000 mg, preferably from about 50 to about 1000 mg, more preferably from about 100 to 750 mg, and most preferably from about 200 to about 500 mg.
  • this invention encompasses a method of reducing the time between therapeutically safe and effective doses of irinotecan. Consequently, in one specific embodiment of the invention, irinotecan is administered in a cycle of less than about three weeks (e.g., about once every two weeks, about once every ten days, or about once every week). The invention further allows the frequency, number, and length of irinotecan dosing cycles to be increased. Thus, another specific embodiment of the invention encompasses the administration of irinotecan for more cycles than are typical when it is administered alone. See, e.g., Physicians' Desk Reference, 2412-2418 (54 th ed., 2000).
  • irinotecan is administered for a greater number of cycles that would typically cause dose-limiting toxicity in a patient to whom thalidomide is not also being administered.
  • irinotecan is administered by intravenous infusion over about 90 minutes every cycle.
  • one cycle comprises the administration of about 125 mg/m 2 irinotecan on days 1, 8, 15, and 22, and then two weeks of rest.
  • each cycles comprises the administration of about 350 mg/m 2 of irinotecan, followed by three weeks of rest.
  • the number of cycles during which irinotecan is administered to a patient will be from about 1 to about 12 cycles, more typically from about 2 to about 10 cycles, and even more typically from about 2 to about 8 cycles.
  • dosage amounts and frequencies provided above are encompassed by the terms “therapeutically effective,” “prophylactically effective,” and “therapeutically or prophylactically effective” as used herein.
  • therapeutically effective When used in connection with an amount of a thalidomide or thalidomide derivative, these terms further encompass an amount of thalidomide or thalidomide derivative that reduces, prevents, or eliminates an adverse effect associated with the administration of irinotecan.
  • This invention encompasses a method of increasing the dosage of irinotecan that can be safely and effectively administered to a patient.
  • This method comprises administering to a patient (e.g., a human) thalidomide, or a pharmaceutically acceptable derivative, salt, solvate, clathrate, hydrate, or prodrug thereof.
  • Patients that can benefit by this method are those likely to suffer from an adverse effect associated with irinotecan that is alleviated or reduced by the administration of thalidomide, or a pharmaceutically acceptable derivative, salt, solvate, clathrate, hydrate, or prodrug thereof, and which is of such severity that it would otherwise limit the amount of irinotecan that can be safely and effectively administered to them.
  • adverse effects are referred to herein as "dose-limiting.”
  • adverse effects that are associated with irinotecan and which can limit the amount of irinotecan that can safely and effectively be administered to a patient include, but are not limited to, early and late-forming diarrhea, nausea, vomiting, anorexia, constipation, flatulence, leukopenia, anemia, neutropenia, asthenia, abdominal cramping, fever, pain, loss of body weight, dehydration, alopecia, dyspnea, insomnia, and dizziness.
  • thalidomide, or a pharmaceutically acceptable derivative, salt, solvate, clathrate, hydrate,.or prodrug thereof is administered prior to, during, or after irinotecan.
  • thalidomide is administered orally and daily in an amount of from about 1 to about 2000 mg, preferably from about 50 to about 1000 mg, more preferably from about 100 to 750 mg, and most preferably from about 200 to about 500 mg.
  • PHARMACEUTICAL COMPOSITIONS AND DOSAGE FORMS is administered orally and daily in an amount of from about 1 to about 2000 mg, preferably from about 50 to about 1000 mg, more preferably from about 100 to 750 mg, and most preferably from about 200 to about 500 mg.
  • compositions can be used in the preparation of individual dosage forms. Consequently, pharmaceutical compositions and dosage forms of the invention comprise the active ingredients disclosed herein (i.e., irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, and thalidomide, a derivative or analogue of thalidomide, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof). Pharmaceutical compositions and dosage forms of the invention can further comprise one or more excipients.
  • active ingredients disclosed herein i.e., irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof
  • thalidomide a derivative or analogue of thalidomide
  • Pharmaceutical compositions and dosage forms of the invention can further comprise one or more excipients.
  • Single unit dosage forms of the invention are suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial), or transdermal administration to a patient.
  • mucosal e.g., nasal, sublingual, vaginal, buccal, or rectal
  • parenteral e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial
  • transdermal administration to a patient.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amo ⁇ hous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • suspensions e.g., aque
  • compositions, shape, and type of dosage forms of the invention will typically vary depending on their use.
  • a dosage form used in the acute treatment of a disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease.
  • Suitable excipients are well known to those skilled in the art of.pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for inco ⁇ oration into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient.
  • oral dosage forms such as tablets may contain excipients not suited for use in parenteral dosage forms.
  • the suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients may be accelerated by some excipients such as lactose, or when exposed to water. Active ingredients that comprise primary or secondary amines are particularly susceptible to such accelerated decomposition.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • the amounts and specific types of active ingredients in a dosage form may differ depending on factors such as, but not limited to, the route by which it is to be administered to patients.
  • typical dosage forms of the invention comprise thalidomide, a derivative or analogue of thalidomide, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, or prodrug thereof in an amount of from about 1 mg to about 2000 mg, more preferably from about 50 mg to about 1000 mg, even more preferably from about 100 mg to about 750 mg, and most preferably from about 200 mg to about 500 mg.
  • typical dosage forms of the invention comprise irinotecan or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, prodrug or derivative thereof in an amount of from about 1 mg to about 1000 mg, more preferably from about 25 mg to about 750 mg, even more preferably from about 50 mg to about 500 mg, and most preferably from about 100 mg to about 350 mg.
  • compositions of the invention that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
  • Typical oral dosage forms of the invention are prepared by combining the active ingredients in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos.
  • microcrystalline cellulose and mixtures thereof.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101, AVICEL-PH-103 AVICEL RC-581, AVICEL-PH-105 (available from FMC Co ⁇ oration, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof.
  • An specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL-PH-103TM and Starch 1500 LM.
  • fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions of the invention is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Disintegrants are used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention.
  • the amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, preferably from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • calcium stearate e.g., magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc
  • hydrogenated vegetable oil e.g., peanut oil, cottonseed oil
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are inco ⁇ orated.
  • AEROSIL 200 a syloid silica gel
  • a coagulated aerosol of synthetic silica marketed by Degussa Co. of Piano, TX
  • CAB-O-SIL a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA
  • lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are inco ⁇ orated.
  • a preferred solid oral dosage form of the invention comprises thalidomide, anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
  • Active ingredients of the invention can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, 5,674,533, 5,059,595, 5,591,767, 5,120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566, each of which is inco ⁇ orated herein by reference.
  • Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients of the invention.
  • the invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
  • controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counte ⁇ arts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time.
  • the drug In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled- release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous, bolus injection, intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art.
  • Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil,. ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glyco
  • cyclodextrin and its derivatives can be used to increase the solubility of thalidomide and its derivatives. See, e.g., U.S. Patent No. 5,134,127, which is inco ⁇ orated herein by reference.
  • a preferred parenteral composition of the invention is intended for dilution with 5%
  • Dextrose Injection USP
  • 0.9% Sodium Chloride Injection USP
  • Transdermal, topical, and mucosal dosage forms of the invention include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton PA (1980 & 1990); and Introduction to Pharmaceutical Dosage Forms, ' 4th ed., Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels. Further, transdermal dosage forms include "reservoir type" or "matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients e.g., carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane-l,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are non-toxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art.
  • tissue to be treated additional components may be used prior to, in conjunction with, or subsequent to treatment with active ingredients of the invention.
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (P ' ovidone, Polyvidone); urea; and various water-soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and
  • Span 60 (sorbitan monostearate).
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • active ingredients of the invention are preferably not administered to a patient at the same time or by the same route of administration.
  • This invention therefore encompasses kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a patient.
  • a typical kit of the invention comprises a dosage form of irinotecan, or a pharmaceutically acceptable prodrug, salt, solvate, hydrate, or clathrate thereof, -and a dosage form of thalidomide, or a pharmaceutically acceptable derivative, prodrug, salt, solvate, hydrate, or clathrate thereof.
  • Kits of the invention can further comprise devices that are used to administer the active ingredients.
  • devices include, but are not limited to, syringes, drip bags, patches, and inhalers.
  • Kits of the invention can further comprise pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients.
  • the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration.
  • Examples of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water-miscible vehicles such as, but not limited to, ethyl alcohol
  • a specific kit of the invention comprises a solid dosage form of thalidomide suitable for oral administration to a patient, and a liquid dosage form of irinotecan suitable for dilution and parenteral administration to a patient.
  • a preferred oral dosage form of thalidomide comprises 50 mg thalidomide, anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
  • a preferred liquid dosage form of irinotecan comprises 100 mg irinotecan hydrochloride, sorbitol NF powder, and lactic acid, USP, and has a pH of from about 3.0 to about 3.8.
  • the p-values shown in Table 1 refer to the probability of observing the specified number of Grade 1-4 symptoms among 9 patients, given the expected frequencies of Grade 1-4 toxicity. Expected frequencies of toxicity were obtained from Rothenberg, M.L., et al, J. Clin. Oncology 14(4): 1128-1135 (1996); Pitot, H.C., et al, J. Clin. Oncology 15(8):2910- 2919 (1997); and Rothenberg, M.L., et al, Cancer 85(4):786-795 (1999).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Plural Heterocyclic Compounds (AREA)
PCT/US2001/015326 2000-05-15 2001-05-10 Compositions and methods for the treatment of colorectal cancer WO2001087306A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
NZ522766A NZ522766A (en) 2000-05-15 2001-05-10 Compositions and methods for the treatment of colorectal cancer with irinotecan and thalidomide to reduce side effects
DE60118547T DE60118547T2 (de) 2000-05-15 2001-05-10 Pharmazeutische zusammensetzung zur behandlung von kolorektalem krebs welche thalidomid und irinotecan enthält
AU2001261473A AU2001261473B2 (en) 2000-05-15 2001-05-10 Compositions and methods for the treatment of colorectal cancer
EP01935372A EP1286671B1 (en) 2000-05-15 2001-05-10 Compositions for the treatment of colorectal cancer comprising thalidomide and irinotecan
CA2408710A CA2408710C (en) 2000-05-15 2001-05-10 Compositions and methods for the treatment of colorectal cancer
AU6147301A AU6147301A (en) 2000-05-15 2001-05-10 Compositions and methods for the treatment of colorectal cancer
JP2001583774A JP4679028B2 (ja) 2000-05-15 2001-05-10 結腸直腸癌を治療するための組成物および方法
HK03106060A HK1053975A1 (en) 2000-05-15 2003-08-23 Compositions for the treatment of colorectal cancer comprising thalidomide and irinotecan

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US20414200P 2000-05-15 2000-05-15
US60/204,142 2000-05-15

Publications (2)

Publication Number Publication Date
WO2001087306A2 true WO2001087306A2 (en) 2001-11-22
WO2001087306A8 WO2001087306A8 (en) 2002-10-03

Family

ID=22756801

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/015326 WO2001087306A2 (en) 2000-05-15 2001-05-10 Compositions and methods for the treatment of colorectal cancer

Country Status (13)

Country Link
US (2) US6914067B2 (es)
EP (1) EP1286671B1 (es)
JP (1) JP4679028B2 (es)
AT (1) ATE322265T1 (es)
AU (2) AU2001261473B2 (es)
CA (1) CA2408710C (es)
DE (1) DE60118547T2 (es)
DK (1) DK1286671T3 (es)
ES (1) ES2261415T3 (es)
HK (1) HK1053975A1 (es)
NZ (1) NZ522766A (es)
PT (1) PT1286671E (es)
WO (1) WO2001087306A2 (es)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003013533A2 (en) * 2001-07-23 2003-02-20 Epidauros Biotechnologie Ag Methods for improved treatment of cancer with irinotecan based on mrp1
US7189740B2 (en) 2002-10-15 2007-03-13 Celgene Corporation Methods of using 3-(4-amino-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione for the treatment and management of myelodysplastic syndromes
EP1900369A1 (en) 2002-10-15 2008-03-19 Celgene Corporation Methods of using and compositions comprising immunomodulatory compounds for the treatment and management of myelodysplastic syndromes
US9056120B2 (en) 2002-10-15 2015-06-16 Celgene Corporation Methods of treating myelodysplastic syndromes with a combination therapy using lenalidomide and azacitidine
US10034872B2 (en) 2014-08-22 2018-07-31 Celgene Corporation Methods of treating multiple myeloma with immunomodulatory compounds in combination with antibodies
US11116782B2 (en) 2002-10-15 2021-09-14 Celgene Corporation Methods of treating myelodysplastic syndromes with a combination therapy using lenalidomide and azacitidine

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6881420B2 (en) * 2000-06-23 2005-04-19 Teva Pharmaceutical Industries Ltd. Compositions and dosage forms for gastric delivery of irinotecan and methods of treatment that use it to inhibit cancer cell proliferation
US7230012B2 (en) * 2002-11-14 2007-06-12 Celgene Corporation Pharmaceutical compositions and dosage forms of thalidomide
US9006267B2 (en) * 2002-11-14 2015-04-14 Celgene Corporation Pharmaceutical compositions and dosage forms of thalidomide
US6987012B2 (en) * 2003-06-11 2006-01-17 Ultra Biotech Limited Biological compositions and methods for treatment of colorectal cancer
AP2006003637A0 (en) * 2003-11-06 2006-06-30 Celgene Corp Methods and compositions using thalidomide for thetreatment and management of cancers and other diseases
US8658203B2 (en) 2004-05-03 2014-02-25 Merrimack Pharmaceuticals, Inc. Liposomes useful for drug delivery to the brain
EP1746976B1 (en) 2004-05-03 2017-01-11 Merrimack Pharmaceuticals, Inc. Liposomes useful for drug delivery
CA2503099A1 (en) * 2005-04-18 2006-10-18 Kabushiki Kaisha Yakult Honsha Pharmaceutical compositions containing camptothecins
CN101180061A (zh) * 2005-05-19 2008-05-14 普罗米蒂克生物科学公司 化合物、含有此类化合物的组合物、和治疗转移性黑素瘤和其它癌症的方法
KR20080066663A (ko) * 2005-09-21 2008-07-16 씨씨씨 디아그노스틱스 엘엘씨 맞춤화된 항암 화학요법(pac)을 위한 종합적인 진단테스트 방법
US20070219825A1 (en) * 2005-11-23 2007-09-20 Maetzold Derek J Method of managing and reducing side effects associated with exposure to a drug
US7723301B2 (en) * 2007-08-29 2010-05-25 The Board Of Trustees Of The University Of Arkansas Pharmaceutical compositions comprising an anti-teratogenic compound and applications of the same
US20090137631A1 (en) * 2007-11-22 2009-05-28 National Yang-Ming University Methods and pharmaceutical compositions for regulation of g- and/or gc-rich nucleic acid expression
US8709419B2 (en) 2010-08-17 2014-04-29 Hoffmann-La Roche, Inc. Combination therapy
US20120045433A1 (en) * 2010-08-17 2012-02-23 Kapil Dhingra Combination therapy
US9295669B2 (en) 2010-12-14 2016-03-29 Hoffman La-Roche Inc. Combination therapy for proliferative disorders
US9717724B2 (en) 2012-06-13 2017-08-01 Ipsen Biopharm Ltd. Methods for treating pancreatic cancer using combination therapies
AU2013202947B2 (en) 2012-06-13 2016-06-02 Ipsen Biopharm Ltd. Methods for treating pancreatic cancer using combination therapies comprising liposomal irinotecan
US10098813B2 (en) 2014-09-03 2018-10-16 Sun Pharmaceutical Industries Limited Perfusion dosage form
US11318131B2 (en) 2015-05-18 2022-05-03 Ipsen Biopharm Ltd. Nanoliposomal irinotecan for use in treating small cell lung cancer
EP3337467B1 (en) 2015-08-20 2020-12-09 Ipsen Biopharm Ltd. Combination therapy using liposomal irinotecan and a parp inhibitor for cancer treatment
AU2016310476B8 (en) 2015-08-21 2021-10-07 Ipsen Biopharm Ltd. Methods for treating metastatic pancreatic cancer using combination therapies comprising liposomal irinotecan and oxaliplatin
MA42991A (fr) 2015-10-16 2018-08-22 Ipsen Biopharm Ltd Stabilisation de compositions pharmaceutiques de camptothécine
ES2968415T3 (es) 2016-02-09 2024-05-09 Sun Pharmaceutical Ind Ltd Sistema de perfusión
JP2019533684A (ja) 2016-11-02 2019-11-21 イプセン バイオファーム リミティド リポソームイリノテカン、オキサリプラチン、5−フルオロウラシル(およびロイコボリン)を含む併用療法を用いる胃がんの処置
US10537585B2 (en) 2017-12-18 2020-01-21 Dexcel Pharma Technologies Ltd. Compositions comprising dexamethasone
CN116333154A (zh) * 2019-08-19 2023-06-27 美勒斯公司 用结合lgr5和egfr的抗体与拓扑异构酶i抑制剂的组合治疗癌症

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6019790A (ja) 1983-07-14 1985-01-31 Yakult Honsha Co Ltd 新規なカンプトテシン誘導体
AU1531492A (en) 1991-02-14 1992-09-15 Rockefeller University, The Method for controlling abnormal concentration tnf alpha in human tissues
US5629327A (en) 1993-03-01 1997-05-13 Childrens Hospital Medical Center Corp. Methods and compositions for inhibition of angiogenesis
US5643915A (en) 1995-06-06 1997-07-01 Andrulis Pharmaceuticals Corp. Treatment of ischemia/reperfusion injury with thalidomide alone or in combination with other therapies
US5654312A (en) 1995-06-07 1997-08-05 Andrulis Pharmaceuticals Treatment of inflammatory and/or autoimmune dermatoses with thalidomide alone or in combination with other agents
US5955466A (en) * 1997-02-27 1999-09-21 Pharmacia & Upjohn Company Tamoxifen as a therapy to reduce irinotecan hydrochloride-induced diarrhea

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
No Search *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003013533A3 (en) * 2001-07-23 2003-10-09 Epidauros Biotechnologie Ag Methods for improved treatment of cancer with irinotecan based on mrp1
WO2003013536A3 (en) * 2001-07-23 2003-12-18 Epidauros Biotechnologie Ag Methods for treatment of cancer using irinotecan based on ugt1a1
WO2003013533A2 (en) * 2001-07-23 2003-02-20 Epidauros Biotechnologie Ag Methods for improved treatment of cancer with irinotecan based on mrp1
WO2003013535A2 (en) * 2001-07-23 2003-02-20 Epidauros Biotechnologie Ag Use of irinotecan for improved treatment of cancer based on mdr1
WO2003013536A2 (en) * 2001-07-23 2003-02-20 Epidauros Biotechnologie Ag Methods for treatment of cancer using irinotecan based on ugt1a1
WO2003013535A3 (en) * 2001-07-23 2003-09-25 Epidauros Biotechnologie Ag Use of irinotecan for improved treatment of cancer based on mdr1
WO2003013537A3 (en) * 2001-07-23 2003-09-25 Epidauros Biotechnologie Ag Irinotecan for treatment of cancer
WO2003013534A3 (en) * 2001-07-23 2003-10-09 Epidauros Biotechnologie Ag Methods for the treatment of cancer with irinotecan based on cyp3a5
WO2003013534A2 (en) * 2001-07-23 2003-02-20 Epidauros Biotechnologie Ag Methods for the treatment of cancer with irinotecan based on cyp3a5
WO2003013537A2 (en) * 2001-07-23 2003-02-20 Epidauros Biotechnologie Ag Irinotecan for treatment of cancer
US11116782B2 (en) 2002-10-15 2021-09-14 Celgene Corporation Methods of treating myelodysplastic syndromes with a combination therapy using lenalidomide and azacitidine
US7189740B2 (en) 2002-10-15 2007-03-13 Celgene Corporation Methods of using 3-(4-amino-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione for the treatment and management of myelodysplastic syndromes
US7393863B2 (en) 2002-10-15 2008-07-01 Celgene Corporation Methods of using N-{[2-(2,6-dioxo(3-piperidyl)-1,3-dioxoisoindolin-4-yl]methyl}cyclopropyl-carboxamide for the treatment and management of myelodysplastic syndromes
US7863297B2 (en) 2002-10-15 2011-01-04 Celgene Corporation Methods of using 4-(amino)-2-(2,6-dioxo(3-piperidly))-isoindoline-3-dione for the treatment of myelodysplastic syndromes
US9056120B2 (en) 2002-10-15 2015-06-16 Celgene Corporation Methods of treating myelodysplastic syndromes with a combination therapy using lenalidomide and azacitidine
US9925207B2 (en) 2002-10-15 2018-03-27 Celgene Corporation Methods of treating myelodysplastic syndromes using lenalidomide
EP1900369A1 (en) 2002-10-15 2008-03-19 Celgene Corporation Methods of using and compositions comprising immunomodulatory compounds for the treatment and management of myelodysplastic syndromes
EP3219317A1 (en) 2010-05-11 2017-09-20 Celgene Corporation Methods of treating myelodysplastic syndromes with a combination therapy using lenalidomide and azacitidine
US10034872B2 (en) 2014-08-22 2018-07-31 Celgene Corporation Methods of treating multiple myeloma with immunomodulatory compounds in combination with antibodies

Also Published As

Publication number Publication date
US6914067B2 (en) 2005-07-05
WO2001087306A8 (en) 2002-10-03
DK1286671T3 (da) 2006-07-17
US20020035091A1 (en) 2002-03-21
EP1286671A2 (en) 2003-03-05
PT1286671E (pt) 2006-08-31
JP4679028B2 (ja) 2011-04-27
EP1286671B1 (en) 2006-04-05
JP2003533483A (ja) 2003-11-11
DE60118547T2 (de) 2007-02-01
AU2001261473B2 (en) 2006-09-14
CA2408710A1 (en) 2001-11-22
US20050203060A1 (en) 2005-09-15
US7479499B2 (en) 2009-01-20
HK1053975A1 (en) 2003-11-14
ATE322265T1 (de) 2006-04-15
NZ522766A (en) 2005-09-30
ES2261415T3 (es) 2006-11-16
DE60118547D1 (de) 2006-05-18
CA2408710C (en) 2010-01-26
AU6147301A (en) 2001-11-26

Similar Documents

Publication Publication Date Title
US7479499B2 (en) Methods of using thalidomide in combination with irinotecan
AU2001261473A1 (en) Compositions and methods for the treatment of colorectal cancer
EP1505973B1 (en) Combinations for treating multiple myeloma
DE60118571T2 (de) Pharmazeutische zusammensetzungen zur behandlung von krebs welche thalidomid und topoisomerase inhibitoren enthalten
EP1487461B1 (en) Methods of using and compositions comprising immunomodulatory compounds for the treatment and management of myelodysplastic syndromes
US20010012850A1 (en) Methods and compositions for inhibition of angiogenesis
US20080213219A1 (en) Methods for treatment and management of macular degeneration using 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione
AU2001261474A1 (en) Compositions and methods for the treatment of cancer
CA2563748A1 (en) Methods of using and compositions comprising thalidomide for the treatment and management of pulmonary hypertension
WO2004041190A2 (en) Composition for the treatment of macular degenration
WO2008066783A2 (en) Therapeutic materials and methods
NZ547127A (en) Methods and compositions comprising thalidomide for treatment of fibromyalgia
AU2003285107B2 (en) Methods of using and compositions comprising selective cytokine inhibitory drugs for treatment and management of macular degeneration
NZ548537A (en) Methods and compositions using thalidomide for the treatment and management of central nervous system disorders or diseases
EP1676577A1 (en) Compositions for the treatment of cancer comprising a topoisomerase inhibitor and thalidomide
Vet—QC02CA04 Doxazosin Mesilate (BANM, rINNM)
MXPA06004627A (es) Metodos para usar y composiciones que comprenden compuestos inmunomoduladores para tratamiento y manejo de degeneracion macular

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: C1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

D17 Declaration under article 17(2)a
WWE Wipo information: entry into national phase

Ref document number: 2408710

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2001261473

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 522766

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2001935372

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001935372

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 522766

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 522766

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 2001935372

Country of ref document: EP